Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Cell ; 78(2): 329-345.e9, 2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32268122

RESUMO

Neural stem and progenitor cells (NSPCs) are critical for continued cellular replacement in the adult brain. Lifelong maintenance of a functional NSPC pool necessitates stringent mechanisms to preserve a pristine proteome. We find that the NSPC chaperone network robustly maintains misfolded protein solubility and stress resilience through high levels of the ATP-dependent chaperonin TRiC/CCT. Strikingly, NSPC differentiation rewires the cellular chaperone network, reducing TRiC/CCT levels and inducing those of the ATP-independent small heat shock proteins (sHSPs). This switches the proteostasis strategy in neural progeny cells to promote sequestration of misfolded proteins into protective inclusions. The chaperone network of NSPCs is more effective than that of differentiated cells, leading to improved management of proteotoxic stress and amyloidogenic proteins. However, NSPC proteostasis is impaired by brain aging. The less efficient chaperone network of differentiated neural progeny may contribute to their enhanced susceptibility to neurodegenerative diseases characterized by aberrant protein misfolding and aggregation.


Assuntos
Envelhecimento/genética , Chaperonas Moleculares/genética , Células-Tronco Neurais/metabolismo , Agregação Patológica de Proteínas/genética , Trifosfato de Adenosina/genética , Envelhecimento/patologia , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Diferenciação Celular/genética , Chaperoninas/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Redes Reguladoras de Genes/genética , Camundongos , Chaperonas Moleculares/metabolismo , Células-Tronco Neurais/patologia , Dobramento de Proteína , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Células-Tronco/metabolismo , Células-Tronco/patologia
2.
Methods Mol Biol ; 1873: 241-251, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30341614

RESUMO

Fluorescence-based nanoscopy methods (also known as "superresolution" microscopy) have substantially expanded our options to examine the distributions of molecules inside cells with nanometer-scale resolution and molecular specificity. In the biophysical analysis of aggregation-prone misfolded proteins and peptides, this has enabled the visualization of distinct populations of aggregated species such as fibrillar assemblies within intact neuronal cells, well below previous limits of sensitivity and resolution. With the Huntington's disease protein, polyglutamine-expanded mutant huntingtin, as an example, we provide sample preparation and imaging protocols for superresolution microscopy down to the ~30 nm-level.


Assuntos
Imunofluorescência , Proteína Huntingtina/metabolismo , Microscopia de Fluorescência , Proteínas Mutantes , Agregados Proteicos , Agregação Patológica de Proteínas/metabolismo , Animais , Análise de Dados , Proteína Huntingtina/química , Proteína Huntingtina/genética , Neurônios/metabolismo , Neurônios/patologia , Células PC12 , Agregação Patológica de Proteínas/genética , Ratos
3.
PLoS One ; 9(4): e92408, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24691167

RESUMO

The Copper Metabolism MURR1 domain protein 1 (COMMD1) is a protein involved in multiple cellular pathways, including copper homeostasis, NF-κB and hypoxia signalling. Acting as a scaffold protein, COMMD1 mediates the levels, stability and proteolysis of its substrates (e.g. the copper-transporters ATP7B and ATP7A, RELA and HIF-1α). Recently, we established an interaction between the Cu/Zn superoxide dismutase 1 (SOD1) and COMMD1, resulting in a decreased maturation and activation of SOD1. Mutations in SOD1, associated with the progressive neurodegenerative disorder Amyotrophic Lateral Sclerosis (ALS), cause misfolding and aggregation of the mutant SOD1 (mSOD1) protein. Here, we identify COMMD1 as a novel regulator of misfolded protein aggregation as it enhances the formation of mSOD1 aggregates upon binding. Interestingly, COMMD1 co-localizes to the sites of mSOD1 inclusions and forms high molecular weight complexes in the presence of mSOD1. The effect of COMMD1 on protein aggregation is client-specific as, in contrast to mSOD1, COMMD1 decreases the abundance of mutant Parkin inclusions, associated with Parkinson's disease. Aggregation of a polyglutamine-expanded Huntingtin, causative of Huntington's disease, appears unaltered by COMMD1. Altogether, this study offers new research directions to expand our current knowledge on the mechanisms underlying aggregation disease pathologies.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Agregados Proteicos , Dobramento de Proteína , Esclerose Lateral Amiotrófica/metabolismo , Animais , Células HEK293 , Células HeLa , Humanos , Camundongos , Peso Molecular , Proteínas Mutantes/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica , Multimerização Proteica , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo
4.
Curr Opin Cell Biol ; 26: 139-146, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24463332

RESUMO

Failure to maintain protein homeostasis is associated with aggregation and cell death, and underies a growing list of pathologies including neurodegenerative diseases, aging, and cancer. Misfolded proteins can be toxic and interfere with normal cellular functions, particularly during proteotoxic stress. Accordingly, molecular chaperones, the ubiquitin-proteasome system (UPS) and autophagy together promote refolding or clearance of misfolded proteins. Here we discuss emerging evidence that the pathways of protein quality control (PQC) are intimately linked to cell architecture, and sequester proteins into spatially and functionally distinct PQC compartments. This sequestration serves a number of functions, including enhancing the efficiency of quality control; clearing the cellular milieu of potentially toxic species and facilitating asymmetric inheritance of damaged proteins to promote rejuvenation of daughter cells.


Assuntos
Proteínas/metabolismo , Animais , Ciclo Celular , Humanos , Dobramento de Proteína , Transporte Proteico
5.
Cell Mol Life Sci ; 69(1): 149-63, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21667063

RESUMO

Menkes disease (MD) is an X-linked recessive disorder characterized by copper deficiency resulting in a diminished function of copper-dependent enzymes. Most MD patients die in early childhood, although mild forms of MD have also been described. A diversity of mutations in the gene encoding of the Golgi-resident copper-transporting P(1B)-type ATPase ATP7A underlies MD. To elucidate the molecular consequences of the ATP7A mutations, various mutations in ATP7A associated with distinct phenotypes of MD (L873R, C1000R, N1304S, and A1362D) were analyzed in detail. All mutants studied displayed changes in protein expression and intracellular localization parallel to a dramatic decline in their copper-transporting capacity compared to ATP7A the wild-type. We restored these observed defects in ATP7A mutant proteins by culturing the cells at 30°C, which improves the quality of protein folding, similar to that which as has recently has been demonstrated for misfolded ATP7B, a copper transporter homologous to ATP7A. Further, the effect of the canine copper toxicosis protein COMMD1 on ATP7A function was examined as COMMD1 has been shown to regulate the proteolysis of ATP7B proteins. Interestingly, in addition to adjusted growth temperature, binding of COMMD1 partially restored the expression, subcellular localization, and copper-exporting activities of the ATP7A mutants. However, no effect of pharmacological chaperones was observed. Together, the presented data might provide a new direction for developing therapies to improve the residual exporting activity of unstable ATP7A mutant proteins, and suggests a potential role for COMMD1 in this process.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenosina Trifosfatases , Proteínas de Transporte de Cátions , Síndrome dos Cabelos Torcidos , Adenosina Trifosfatases/deficiência , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Proteínas de Transporte de Cátions/deficiência , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Técnicas de Cultura de Células/métodos , Linhagem Celular Tumoral , Cobre/deficiência , Cobre/metabolismo , ATPases Transportadoras de Cobre , Cães , Complexo de Golgi/metabolismo , Células HEK293 , Humanos , Síndrome dos Cabelos Torcidos/genética , Síndrome dos Cabelos Torcidos/metabolismo , Camundongos , Mutação/genética , Dobramento de Proteína , Transporte Proteico/genética , Proteólise , Temperatura
6.
J Biol Chem ; 285(37): 28991-9000, 2010 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-20595380

RESUMO

The maturation and activation of the anti-oxidant Cu,Zn superoxide dismutase (SOD1) are highly regulated processes that require several post-translational modifications. The maturation of SOD1 is initiated by incorporation of zinc and copper ions followed by disulfide oxidation leading to the formation of enzymatically active homodimers. Our present data indicate that homodimer formation is a regulated final step in SOD1 maturation and implicate the recently characterized copper homeostasis protein COMMD1 in this process. COMMD1 interacts with SOD1, and this interaction requires CCS-mediated copper incorporation into SOD1. COMMD1 does not regulate disulfide oxidation of SOD1 but reduces the level of SOD1 homodimers. RNAi-mediated knockdown of COMMD1 expression results in a significant induction of SOD1 activity and a consequent decrease in superoxide anion concentrations, whereas overexpression of COMMD1 exerts exactly the opposite effects. Here, we identify COMMD1 as a novel protein regulating SOD1 activation and associate COMMD1 function with the production of free radicals.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas/metabolismo , Superóxido Dismutase/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/genética , Cobre/metabolismo , Ativação Enzimática/fisiologia , Técnicas de Silenciamento de Genes , Células Hep G2 , Humanos , Camundongos , Proteínas/genética , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Superóxidos/metabolismo
7.
Traffic ; 10(5): 514-27, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19220812

RESUMO

Copper metabolism MURR1 domain1 (COMMD1) is a novel inhibitor of the transcription factors NF-kappaB and HIF-1, which play important roles in inflammation and tumor growth, respectively. In this study, we identified two highly conserved nuclear export signals (NESs) in COMMD1 and revealed that these NESs were essential and sufficient to induce maximal nuclear export of COMMD1. Inhibition of CRM1-mediated nuclear export by Leptomycin B resulted in nuclear accumulation of COMMD1. In addition, low oxygen concentrations induced the active export of COMMD1 from the nucleus in a CRM1-dependent manner. Disruption of the NESs in COMMD1 increased the repression of COMMD1 in transcriptional activity of NF-kappaB and HIF-1. In conclusion, these data indicate that COMMD1 undergoes constitutive nucleocytoplasmic transport as a novel mechanism to regulate NF-kappaB and HIF-1 signaling.


Assuntos
Núcleo Celular/metabolismo , Citosol/metabolismo , Fator 1 Induzível por Hipóxia/metabolismo , NF-kappa B/metabolismo , Fatores de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular , Cobre/metabolismo , Ácidos Graxos Insaturados , Humanos , Sinais de Exportação Nuclear , Transdução de Sinais , Fator de Transcrição RelA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA